1
|
Dos Santos L, Cristino de Oliveira A, Marcondes Silva Lotz R, Manera F, Fernandes RC, Moreira Castilho B, Höfelmann DA. Trajectory Patterns of Weight and Length in Children: From Birth Until 2-Years of Age. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2024; 43:686-696. [PMID: 38990649 DOI: 10.1080/27697061.2024.2374412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 06/07/2024] [Accepted: 06/26/2024] [Indexed: 07/13/2024]
Abstract
OBJECTIVE The aim was to identify trajectory patterns of weight and length in children from birth until two years of life and establish associations with maternal and child characteristics. METHODS A mixed-cohort study was conducted in public health services in Colombo-PR, Brazil, between 2018 and 2022. Pregnancy information was gathered through anthropometric data collection and questionnaires. Birth data were extracted from birth record forms, while weight and length data in the first two years of life were obtained from physical and electronic health service records. Weight and length trajectory patterns were identified using a group-based trajectory model. The definition of the number of trajectory patterns to be selected considered the model fit to the type of variable, its practical utility, as well as the probabilities of group membership. RESULTS Two trajectory patterns of weight and length were identified among the children. The majority exhibited a pattern of weight (67.8%, n = 382) and length (90.9%, n = 472) considered high and stable, with a tendency to decelerate from one and a half years of age. The probability of belonging to the lower weight gain group was associated with female sex (41.5%, p < 0.001), smoking during pregnancy (48.7%, p = 0.008), prematurity (65.0%, p = 0.001), cesarean delivery (36.4%, p = 0.009), small for gestational age (69.0%, p < 0.001), and twinning (69.2%, p = 0.002). Similarly, the probability of belonging to the lower length gain group was associated with female sex (11.7%, p < 0.001), smoking during pregnancy (20.6%, p = 0.003), cesarean delivery (10.1%, p = 0.048), born small for gestational age (46.4%, p < 0.001), and twinning (46.1%, p < 0.001). CONCLUSION Conditions during pregnancy and childbirth can impact growth patterns in the first two years of life.
Collapse
Affiliation(s)
- Lais Dos Santos
- Post-Graduate Program of Public Health, Department of Public Health, Federal University of Paraná, Curitiba, Brazil
| | - Andressa Cristino de Oliveira
- Post-Graduate Program of Food and Nutrition, Department of Nutrition, Federal University of Paraná, Curitiba, Brazil
| | - Rafaela Marcondes Silva Lotz
- Post-Graduate Program of Food and Nutrition, Department of Nutrition, Federal University of Paraná, Curitiba, Brazil
| | - Fernanda Manera
- Post-Graduate Program of Food and Nutrition, Department of Nutrition, Federal University of Paraná, Curitiba, Brazil
| | - Renata Cordeiro Fernandes
- Post-Graduate Program of Food and Nutrition, Department of Nutrition, Federal University of Paraná, Curitiba, Brazil
| | - Barbara Moreira Castilho
- Post-Graduate Program of Food and Nutrition, Department of Nutrition, Federal University of Paraná, Curitiba, Brazil
| | - Doroteia Aparecida Höfelmann
- Post-Graduate Program of Public Health, Department of Public Health, Federal University of Paraná, Curitiba, Brazil
- Post-Graduate Program of Food and Nutrition, Department of Nutrition, Federal University of Paraná, Curitiba, Brazil
| |
Collapse
|
2
|
Lin C, Cai X, Li Z, Lv F, Yang W, Ji L. The association between exposure to famine in early life and risks of diabetic complications in adult patients with type two diabetes. J Glob Health 2024; 14:04167. [PMID: 39302069 DOI: 10.7189/jogh.14.04167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024] Open
Abstract
Background In this study, we aimed to assess the associations between early exposure to famine and the risks of diabetic complications in adult patients with type two diabetes. Methods The participants in this study were selected from China National HbA1c Surveillance System (2009-13) and further stratified according to the birth year. The participants born between 1956-59, 1959-61, and 1962-64 were classified as foetal exposed group with 70 852, infant/toddler exposed group with 93 616, and unexposed group with 72 723 participants. The association between exposure to famine in early life and risks of diabetic complications were analysed by logistic regression. We assessed the attributing effects of the interaction between exposure to famine in early life and modifiable risk factors by the multiplicative and additive interactive models. Results After adjustments for sex, famine severity, economic status in adulthood, body mass index, blood pressure, low-density lipoprotein cholesterol, glycated haemoglobin, diabetes duration, and the use of antidiabetic agents, the increased risks of coronary heart disease (odds ratio (OR) = 1.31; 95% CI (confidence interval) = 1.26, 1.36), cerebrovascular disease (OR = 1.32; 95% CI = 1.24, 1.41), and diabetic retinopathy (OR = 1.06; 95% CI = 1.02, 1.10) were observed in patients with early-life exposure to famine. The reduced risk of diabetic kidney disease (OR = 0.94; 95% CI = 0.90, 0.99) was observed in patients with early-life exposure to famine compared with those without famine exposure. The interaction analyses indicated that obesity might exacerbate the increased risk of coronary heart disease (OR = 1.26; 95% CI = 1.22, 1.30), cerebrovascular disease (OR = 1.26; 95% CI = 1.21, 1.32), and diabetic retinopathy associated with early-life exposure to famine (OR = 1.09; 95% CI = 1.06, 1.12) in patients with type two diabetes. Moreover, high economic status in adulthood might also exacerbate the increased risk of coronary heart disease (OR = 1.35; 95% CI = 1.30, 1.40) and cerebrovascular disease (OR = 1.33; 95% CI = 1.23, 1.43) associated with early-life exposure to famine in patients with type two diabetes. Conclusions Early-life exposure to famine in patients with type two diabetes might be associated with increased risks of coronary heart disease, cerebrovascular disease, and diabetic retinopathy but a reduced risk of diabetic kidney disease in adulthood. Obesity and high economic status might further exacerbate the risk of diabetic complications associated with early-life exposure to famine. Improving early-life nutritional status may promote better risk prevention and management of diabetic complications in patients with type two diabetes.
Collapse
|
3
|
Baud O, Zana-Taieb E, Vaiman D. Glucocorticosteroids and bronchopulmonary dysplasia : is epigenetics the missing link? Pediatr Res 2024; 96:291-292. [PMID: 38627592 PMCID: PMC11343698 DOI: 10.1038/s41390-024-03203-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 03/25/2024] [Indexed: 08/25/2024]
Affiliation(s)
- Olivier Baud
- Division of Neonatology and Pediatric Intensive Care, Children's University Hospital of Geneva and University of Geneva, Geneva, Switzerland.
- Inserm U1141, University Paris-Cité, Paris, France.
| | - Elodie Zana-Taieb
- Department of Neonatal Medicine of Port-Royal, Cochin Hospital, FHU PREMA, AP-HP Centre-Université Paris Cité, Paris, France
| | - Daniel Vaiman
- Institut Cochin, U1016 INSERM, CNRS UMR8104, Faculté de Paris, 24 Rue du Faubourg St Jacques, Paris, 75014, France
| |
Collapse
|
4
|
Furness D, Huynh NKT, Kaufmann L, Liu J, Nguyen TBN, Schaefer E, Tan L, Yau CD, Yu Q. Real-world insights on nutritional awareness and behaviors among preconception and pregnant women in three Asia Pacific countries. Front Glob Womens Health 2024; 5:1332555. [PMID: 38813069 PMCID: PMC11135049 DOI: 10.3389/fgwh.2024.1332555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 04/22/2024] [Indexed: 05/31/2024] Open
Abstract
Introduction In many parts of Asia Pacific (APAC), insufficient intake of micronutrients that are important for conception and pregnancy remains a prevalent issue among women of reproductive age. It is crucial to gain insights into women's nutritional awareness and nutrition-related behaviors, as well as how these relate to their health literacy (HL). This understanding can help identify gaps and guide the development of appropriate intervention strategies. However, there appears to be limited relevant data available for the APAC region. We therefore examined nutritional awareness and behaviors among preconception and pregnant women in three APAC countries, and explored how these were related to women's HL. Methods Cross-sectional online surveys were conducted among preconception (i.e., planning to conceive within the next 12 months or currently trying to conceive) and pregnant women in Australia (N = 624), China (N = 600), and Vietnam (N = 300). The survey questionnaire included a validated tool for HL (Newest Vital Sign) and questions to examine awareness and behaviors relating to healthy eating and prenatal supplementation during preconception and pregnancy. Results Despite recommendations for a quality diet complemented by appropriate supplementation during preconception and pregnancy, many respondents in each country were not aware of the specific impact of adequate nutrition during these stages. While many respondents reported changes in their diet to eat more healthily during preconception and pregnancy, a substantial proportion were not taking prenatal supplements. Higher HL was related to greater nutritional awareness and higher use of prenatal supplements. Discussion Our findings suggest that there are gaps in nutritional awareness and practices of many preconception or pregnant women in the three countries. Interventions to improve HL would be valuable to complement conventional knowledge-centric nutrition education, and enhance understanding and empower women to adopt appropriate nutritional practices throughout their preconception/pregnancy journey.
Collapse
Affiliation(s)
- Denise Furness
- Nutritionist and Molecular Geneticist, Your Genes and Nutrition, Doonan, QLD, Australia
| | - Nguyen Khanh Trang Huynh
- Department of Obstetrics and Gynaecology, Pham Ngoc Thach University of Medicine, Ho Chi Minh, Vietnam
| | - Ligaya Kaufmann
- Department of Regulatory, Medical, Safety, Quality & Compliance (RMSQC), Bayer Consumer Care AG, Basel, Switzerland
| | - Jue Liu
- Department of Regulatory, Medical, Safety, Quality & Compliance (RMSQC), Bayer Healthcare Company Limited, Shanghai, China
| | - Thi Bich Ngoc Nguyen
- Department of Regulatory, Medical, Safety, Quality & Compliance (RMSQC), Bayer Vietnam Limited, Bien Hoa, Dong Nai, Vietnam
| | - Ella Schaefer
- Department of Regulatory, Medical, Safety, Quality & Compliance (RMSQC), Bayer Consumer Care AG, Basel, Switzerland
| | - Lucy Tan
- Department of Regulatory, Medical, Safety, Quality & Compliance (RMSQC), Bayer Australia Limited, Pymble, NSW, Australia
| | - Ching Danica Yau
- Department of Regulatory, Medical, Safety, Quality & Compliance (RMSQC), Bayer Healthcare Limited, Consumer Health, Hong Kong, Hong Kong SAR, China
| | - Qi Yu
- Gynecological Endocrinology and Reproductive Medicine Center, Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
5
|
Horner S. Impact of Parent Presence and Engagement on Stress in NICU Infants. Adv Neonatal Care 2024; 24:132-140. [PMID: 38547480 DOI: 10.1097/anc.0000000000001146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
BACKGROUND Infants in neonatal intensive care units (NICUs) are exposed to frequent stressors that impact their neurodevelopmental outcomes. Parent presence and engagement are considered critical to improving infant outcomes, yet associations between cumulative NICU parent presence, engagement, and infant stress are infrequently examined. PURPOSE To examine associations between NICU infant stress and the amount (hours per week) or frequency (days per week) of parent presence and skin-to-skin care (SSC). METHODS A secondary analysis of a data set representing 78 NICU families was conducted. Infant acuity was measured using Neonatal Medical Index (NMI) scores. Parent presence and SSC data were collected from electronic medical records. Infant stress was measured using resting salivary cortisol levels collected at NICU discharge (median = 33 days of life). RESULTS More cumulative SSC was associated with lower discharge cortisol in NICU infants for SSC measured in hours per week (P = .03) or days per week (P = .05). Cumulative parent presence was not significantly associated with infant cortisol at discharge. Hierarchical regression analyses examining timing of parent presence supported a model including admission cortisol, NMI score, and parent presence during weeks 1 to 4 of life for explaining infant stress at discharge (R2 = 0.44, P = .004). Analyses examining timing of SSC supported a model including admission cortisol, NMI score, and frequency of SSC during week 1 for explaining infant stress at discharge (R2 = 0.21, P = .04). IMPLICATIONS FOR PRACTICE AND RESEARCH Early, frequent SSC to mitigate stress in NICU infants was supported. Results suggested that timing of parent presence impacts NICU infant stress; however, additional study is recommended.
Collapse
Affiliation(s)
- Susan Horner
- Author Affiliations: Loyola University Chicago, Chicago, Illinois; and Ann & Robert H. Lurie Children's Hospital, Lombard, Illinois
| |
Collapse
|
6
|
Nanninga EK, Menting MD, van der Hijden EJE, Portrait FRM. Do women living in a deprived neighborhood have higher maternity care costs and worse pregnancy outcomes? A retrospective population-based study. BMC Health Serv Res 2024; 24:360. [PMID: 38509560 PMCID: PMC10956252 DOI: 10.1186/s12913-024-10737-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 02/16/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Living in a deprived neighborhood is associated with poorer health, due to factors such as lower socio-economic status and an adverse lifestyle. There is little insight into whether living in deprived neighborhood is associated with adverse maternity care outcomes and maternity health care costs. We expect women in a deprived neighborhood to experience a more complicated pregnancy, with more secondary obstetric care (as opposed to primary midwifery care) and higher maternity care costs. This study aimed to answer the following research question: to what extent are moment of referral from primary to secondary care, mode of delivery, (extreme or very) preterm delivery and maternity care costs associated with neighborhood deprivation? METHODS This retrospective cohort study used a national Dutch database with healthcare claims processed by health insurers. All pregnancies that started in 2018 were included. The moment of referral from primary to secondary care, mode of delivery, (extreme or very) preterm delivery and maternity care costs were compared between women in deprived and non-deprived neighborhoods. We reported descriptive statistics, and results of ordinal logistic, multinomial and linear regressions to assess whether differences between the two groups exist. RESULTS Women in deprived neighborhoods had higher odds of being referred from primary to secondary care during pregnancy (adjusted OR 1.49, 95%CI 1.41-1.57) and to start their pregnancy in secondary care (adjusted OR 1.55, 95%CI 1.44-1.66). Furthermore, women in deprived neighborhoods had lower odds of assisted delivery than women in non-deprived neighborhoods (adjusted OR 0.73, 95%CI 0.66-0.80), and they had higher odds of a cesarean section (adjusted OR 1.19, 95%CI 1.13-1.25). On average, women in a deprived neighborhood had higher maternity care costs worth 156 euros (95%CI 104-208). CONCLUSION This study showed that living in a deprived neighborhood is associated with more intensive maternal care and higher maternal care costs in the Netherlands. These findings support the needs for greater attention to socio-economic factors in maternity care in the Netherlands.
Collapse
Affiliation(s)
- Eline K Nanninga
- School of Business and Economics Department of Health Sciences, Ethics, Governance and Society, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
- Dutch Healthcare Authority, Newtonlaan 1-41, 3584 BX, Utrecht, The Netherlands
| | - Malou D Menting
- Dutch Healthcare Authority, Newtonlaan 1-41, 3584 BX, Utrecht, The Netherlands.
| | - Eric J E van der Hijden
- School of Business and Economics Department of Health Sciences, Ethics, Governance and Society, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
- Zilveren Kruis Health Insurance , Dellaertweg 1, 2316 WZ, Leiden, The Netherlands
| | - France R M Portrait
- School of Business and Economics Department of Health Sciences, Ethics, Governance and Society, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Acharya L, Garg A, Rai M, Kshetri R, Grewal US, Dhakal P. Novel chimeric antigen receptor targets and constructs for acute lymphoblastic leukemia: Moving beyond CD19. J Investig Med 2024; 72:32-46. [PMID: 37497999 DOI: 10.1177/10815589231191811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Acute lymphoblastic leukemia (ALL) is the second most common acute leukemia in adults with a poor prognosis with relapsed or refractory (R/R) B-cell lineage ALL (B-ALL). Anti-CD19 chimeric antigen receptor (CAR) T-cell therapy has shown excellent response rates in RR B-ALL, but most patients relapse due to poor persistence of CAR T-cell therapy or other tumor-associated escape mechanisms. In addition, anti-CD19 CAR T-cell therapy causes several serious side effects such as cytokine release syndrome and neurotoxicity. In this review, we will discuss novel CAR targets, CAR constructs, and various strategies to boost CARs for the treatment of RR B-ALL. In addition, we discuss a few novel strategies developed to reduce the side effects of CAR.
Collapse
Affiliation(s)
- Luna Acharya
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Alpana Garg
- Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Manoj Rai
- Department of Internal Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Rupesh Kshetri
- Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Udhayvir S Grewal
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Prajwal Dhakal
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| |
Collapse
|
8
|
Bukasa LL, Namiba A, Brown M, Ndu'ngu E, Nangwale M, Letting G, Chirwa P, Thorne C, Tariq S. Setting the research agenda: involving parents in research on children who are HIV-free. J Int AIDS Soc 2023; 26 Suppl 4:e26150. [PMID: 37909217 PMCID: PMC10618900 DOI: 10.1002/jia2.26150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 08/14/2023] [Indexed: 11/02/2023] Open
Abstract
INTRODUCTION There is growing interest in health, developmental and survival outcomes of children who are born HIV-free to women living with HIV (children born HIV-free). To date, the research agenda has been largely determined by researchers, funders and policy makers, with limited involvement of parents, who are key stakeholders. Researchers at UCL Great Ormond Street Institute of Child Health in partnership with community-based organisation 4M Network of Mentor Mothers conducted two workshops with parents in March 2022 to establish research priorities for children born HIV-free, and key considerations for methodological approaches both to research and engagement with the affected communities. DISCUSSION When exploring research on children born HIV-free, we consider the following: what aspects of current research are aligned with women and parents' priorities, what is missing and what approaches would be preferred. A holistic approach to research on children born HIV-free should be prioritised, focussing on a breadth of outcomes and how they intersect. Secondary use of existing data sources should be maximised to facilitate this, with a view of monitoring the long-term effects of fetal antiretroviral drug exposure alongside other key health and developmental outcomes. Involving and engaging with parents, and children where possible, must be at the heart of research design to maximise relevance and impact of findings for the affected communities. Potential barriers to engaging with individuals who were children born HIV-free include parental disclosure and individuals not identifying as a child born HIV-free to a mother living with HIV. Stigma-free language must be incorporated into the vocabulary of researchers and other stakeholders, avoiding reference to exposure; we propose the term "children born HIV-free." CONCLUSIONS Mothers and parents living with HIV should be involved in research about their children born HIV-free and are key in identifying research priorities so that findings may translate into an impact on their children's health and wellbeing. Meaningful involvement of women living with HIV through trusted community partners is an effective mechanism by which to elicit views on research about their children.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Claire Thorne
- UCL Great Ormond Institute of Child Health, London, UK
| | - Shema Tariq
- UCL Institute for Global Health, London, UK
- Mortimer Market Centre, Central and North West London NHS Foundation Trust, London, UK
| |
Collapse
|
9
|
Eichenauer H, Ehlert U. The association between prenatal famine, DNA methylation and mental disorders: a systematic review and meta-analysis. Clin Epigenetics 2023; 15:152. [PMID: 37716973 PMCID: PMC10505322 DOI: 10.1186/s13148-023-01557-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 08/14/2023] [Indexed: 09/18/2023] Open
Abstract
BACKGROUND Undernutrition in pregnant women is an unfavorable environmental condition that can affect the intrauterine development via epigenetic mechanisms and thus have long-lasting detrimental consequences for the mental health of the offspring later in life. One epigenetic mechanism that has been associated with mental disorders and undernutrition is alterations in DNA methylation. The effect of prenatal undernutrition on the mental health of adult offspring can be analyzed through quasi-experimental studies such as famine studies. The present systematic review and meta-analysis aims to analyze the association between prenatal famine exposure, DNA methylation, and mental disorders in adult offspring. We further investigate whether altered DNA methylation as a result of prenatal famine exposure is prospectively linked to mental disorders. METHODS We conducted a systematic search of the databases PubMed and PsycINFO to identify relevant records up to September 2022 on offspring whose mothers experienced famine directly before and/or during pregnancy, examining the impact of prenatal famine exposure on the offspring's DNA methylation and/or mental disorders or symptoms. RESULTS The systematic review showed that adults who were prenatally exposed to famine had an increased risk of schizophrenia and depression. Several studies reported an association between prenatal famine exposure and hyper- or hypomethylation of specific genes. The largest number of studies reported differences in DNA methylation of the IGF2 gene. Altered DNA methylation of the DUSP22 gene mediated the association between prenatal famine exposure and schizophrenia in adult offspring. Meta-analysis confirmed the increased risk of schizophrenia following prenatal famine exposure. For DNA methylation, meta-analysis was not suitable due to different microarrays/data processing approaches and/or unavailable data. CONCLUSION Prenatal famine exposure is associated with an increased risk of mental disorders and DNA methylation changes. The findings suggest that changes in DNA methylation of genes involved in neuronal, neuroendocrine, and immune processes may be a mechanism that promotes the development of mental disorders such as schizophrenia and depression in adult offspring. Such findings are crucial given that undernutrition has risen worldwide, increasing the risk of famine and thus also of negative effects on mental health.
Collapse
Affiliation(s)
- Heike Eichenauer
- Clinical Psychology and Psychotherapy, University of Zurich, Binzmühlestrasse 14, 8050, Zurich, Switzerland
| | - Ulrike Ehlert
- Clinical Psychology and Psychotherapy, University of Zurich, Binzmühlestrasse 14, 8050, Zurich, Switzerland.
| |
Collapse
|
10
|
Provenzi L, Bussu G, Riva V. Editorial: Risk and protective factors, family environment and (a)typical neurodevelopmental outcomes. Front Psychol 2023; 14:1221338. [PMID: 37408973 PMCID: PMC10319110 DOI: 10.3389/fpsyg.2023.1221338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 06/12/2023] [Indexed: 07/07/2023] Open
Affiliation(s)
- Livio Provenzi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Developmental Psychobiology Lab, IRCCS Mondino Foundation, Pavia, Italy
| | - Giorgia Bussu
- Development and Neurodiversity Lab, Developmental Psychology Division, Department of Psychology, Uppsala University, Uppsala, Sweden
| | - Valentina Riva
- Child Psychopathology Unit, Scientific Institute IRCCS E. Medea, Lecco, Italy
| |
Collapse
|
11
|
Meredith C, McKerchar C, Lacey C. Indigenous approaches to perinatal mental health: a systematic review with critical interpretive synthesis. Arch Womens Ment Health 2023; 26:275-293. [PMID: 37002367 PMCID: PMC10191969 DOI: 10.1007/s00737-023-01310-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 03/19/2023] [Indexed: 05/18/2023]
Abstract
Indigenous mothers and birthing parents experience significant inequities during the perinatal period, with mental health distress causing adverse outcomes for mothers/birthing parents and their infants. Limited literature is available to inform our understanding of solutions to these issues, with research primarily focusing on inequities. Our aim was to conduct a systematic review of Indigenous approaches to treatment of perinatal mental health illness. Following the PRISMA guidelines for systematic literature reviews, an electronic search of CINAHL, Medline, PubMed, Embase, APA PsycInfo, OVID Nursing, Scopus, Web of Science, and Google Scholar databases was conducted in January and February 2022 and repeated in June 2022. Twenty-seven studies were included in the final review. A critical interpretive synthesis informed our approach to the systematic review. The work of (Yamane and Helm J Prev 43:167-190, 2022) was drawn upon to differentiate studies and place within a cultural continuum framework. Across the 27 studies, the majority of participants were healthcare workers and other staff. Mothers, birthing parents, and their families were represented in small numbers. Outcomes of interest included a reduction in symptoms, a reduction in high-risk behaviours, and parental engagement/attachment of mothers/birthing parents with their babies. Interventions infrequently reported significant reductions in mental health symptoms, and many included studies focused on qualitative assessments of intervention acceptability or utility. Many studies focused on describing approaches to perinatal mental health distress or considered the perspectives and priorities of families and healthcare workers. More research and evaluation of Indigenous interventions for perinatal mental health illness is required. Future research should be designed to privilege the voices, perspectives, and experiences of Indigenous mothers, birthing parents, and their families. Researchers should ensure that any future studies should arise from the priorities of the Indigenous population being studied and be Indigenous-led and designed.
Collapse
Affiliation(s)
- Cara Meredith
- Māori and Indigenous Health Innovation, University of Otago, Christchurch, New Zealand.
| | | | - Cameron Lacey
- Māori and Indigenous Health Innovation, University of Otago, Christchurch, New Zealand
| |
Collapse
|
12
|
Davis O. Abnormal Chromatin Folding in the Molecular Pathogenesis of Epilepsy and Autism Spectrum Disorder: a Meta-synthesis with Systematic Searching. Mol Neurobiol 2023; 60:768-779. [PMID: 36367658 PMCID: PMC9849311 DOI: 10.1007/s12035-022-03106-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/25/2022] [Indexed: 11/13/2022]
Abstract
How DNA is folded and packaged in nucleosomes is an essential regulator of gene expression. Abnormal patterns of chromatin folding are implicated in a wide range of diseases and disorders, including epilepsy and autism spectrum disorder (ASD). These disorders are thought to have a shared pathogenesis involving an imbalance in the number of excitatory-inhibitory neurons formed during neurodevelopment; however, the underlying pathological mechanism behind this imbalance is poorly understood. Studies are increasingly implicating abnormal chromatin folding in neural stem cells as one of the candidate pathological mechanisms, but no review has yet attempted to summarise the knowledge in this field. This meta-synthesis is a systematic search of all the articles on epilepsy, ASD, and chromatin folding. Its two main objectives were to determine to what extent abnormal chromatin folding is implicated in the pathogenesis of epilepsy and ASD, and secondly how abnormal chromatin folding leads to pathological disease processes. This search produced 22 relevant articles, which together strongly implicate abnormal chromatin folding in the pathogenesis of epilepsy and ASD. A range of mutations and chromosomal structural abnormalities lead to this effect, including single nucleotide polymorphisms, copy number variants, translocations and mutations in chromatin modifying. However, knowledge is much more limited into how abnormal chromatin organisation subsequently causes pathological disease processes, not yet showing, for example, whether it leads to abnormal excitation-inhibitory neuron imbalance in human brain organoids.
Collapse
Affiliation(s)
- Oliver Davis
- grid.5335.00000000121885934Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN UK
| |
Collapse
|
13
|
Gaillard V, Chastant S, England G, Forman O, German AJ, Suchodolski JS, Villaverde C, Chavatte-Palmer P, Péron F. Environmental risk factors in puppies and kittens for developing chronic disorders in adulthood: A call for research on developmental programming. Front Vet Sci 2022; 9:944821. [PMID: 36619947 PMCID: PMC9816871 DOI: 10.3389/fvets.2022.944821] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
Many dogs and cats are affected by chronic diseases that significantly impact their health and welfare and relationships with humans. Some of these diseases can be challenging to treat, and a better understanding of early-life risk factors for diseases occurring in adulthood is key to improving preventive veterinary care and husbandry practices. This article reviews early-life risk factors for obesity and chronic enteropathy, and for chronic behavioral problems, which can also be intractable with life-changing consequences. Aspects of early life in puppies and kittens that can impact the risk of adult disorders include maternal nutrition, establishment of the gut microbiome, maternal behavior, weaning, nutrition during growth, growth rate, socialization with conspecifics and humans, rehoming and neutering. Despite evidence in some species that the disorders reviewed here reflect the developmental origins of health and disease (DOHaD), developmental programming has rarely been studied in dogs and cats. Priorities and strategies to increase knowledge of early-life risk factors and DOHaD in dogs and cats are discussed. Critical windows of development are proposed: preconception, gestation, the suckling period, early growth pre-neutering or pre-puberty, and growth post-neutering or post-puberty to adult size, the durations of which depend upon species and breed. Challenges to DOHaD research in these species include a large number of breeds with wide genetic and phenotypic variability, and the existence of many mixed-breed individuals. Moreover, difficulties in conducting prospective lifelong cohort studies are exacerbated by discontinuity in pet husbandry between breeders and subsequent owners, and by the dispersed nature of pet ownership.
Collapse
Affiliation(s)
- Virginie Gaillard
- Research and Development Center, Royal Canin, Aimargues, France,*Correspondence: Virginie Gaillard ✉
| | - Sylvie Chastant
- NeoCare, Université de Toulouse, Ecole Nationale Vétérinaire de Toulouse (ENVT), Toulouse, France
| | - Gary England
- School of Veterinary Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Oliver Forman
- Wisdom Panel, Kinship, Waltham-on-the-Wolds, Leicestershire, United Kingdom
| | - Alexander J. German
- Institute of Life Course and Medical Sciences of Small Animal Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Jan S. Suchodolski
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
| | | | - Pascale Chavatte-Palmer
- Université Paris-Saclay, Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Institut National de Recherche Pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Biologie de la Reproduction, Environnement, Epigénétique et Développement (BREED), Jouy-en-Josas, France,Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France
| | - Franck Péron
- Research and Development Center, Royal Canin, Aimargues, France
| |
Collapse
|
14
|
Melnik BC, Schmitz G. Milk Exosomal microRNAs: Postnatal Promoters of β Cell Proliferation but Potential Inducers of β Cell De-Differentiation in Adult Life. Int J Mol Sci 2022; 23:ijms231911503. [PMID: 36232796 PMCID: PMC9569743 DOI: 10.3390/ijms231911503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic β cell expansion and functional maturation during the birth-to-weaning period is driven by epigenetic programs primarily triggered by growth factors, hormones, and nutrients provided by human milk. As shown recently, exosomes derived from various origins interact with β cells. This review elucidates the potential role of milk-derived exosomes (MEX) and their microRNAs (miRs) on pancreatic β cell programming during the postnatal period of lactation as well as during continuous cow milk exposure of adult humans to bovine MEX. Mechanistic evidence suggests that MEX miRs stimulate mTORC1/c-MYC-dependent postnatal β cell proliferation and glycolysis, but attenuate β cell differentiation, mitochondrial function, and insulin synthesis and secretion. MEX miR content is negatively affected by maternal obesity, gestational diabetes, psychological stress, caesarean delivery, and is completely absent in infant formula. Weaning-related disappearance of MEX miRs may be the critical event switching β cells from proliferation to TGF-β/AMPK-mediated cell differentiation, whereas continued exposure of adult humans to bovine MEX miRs via intake of pasteurized cow milk may reverse β cell differentiation, promoting β cell de-differentiation. Whereas MEX miR signaling supports postnatal β cell proliferation (diabetes prevention), persistent bovine MEX exposure after the lactation period may de-differentiate β cells back to the postnatal phenotype (diabetes induction).
Collapse
Affiliation(s)
- Bodo C. Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany
- Correspondence: ; Tel.: +49-52-4198-8060
| | - Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, University of Regensburg, D-93053 Regensburg, Germany
| |
Collapse
|
15
|
Desale H, Buekens P, Alger J, Cafferata ML, Harville EW, Herrera C, Truyens C, Dumonteil E. Epigenetic signature of exposure to maternal Trypanosoma cruzi infection in cord blood cells from uninfected newborns. Epigenomics 2022; 14:913-927. [PMID: 36039408 PMCID: PMC9475499 DOI: 10.2217/epi-2022-0153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aims: To assess the epigenetic effects of in utero exposure to maternal Trypanosoma cruzi infection. Methods: We performed an epigenome-wide association study to compare the DNA methylation patterns of umbilical cord blood cells from uninfected babies from chagasic and uninfected mothers. DNA methylation was measured using Infinium EPIC arrays. Results: We identified a differential DNA methylation signature of fetal exposure to maternal T. cruzi infection, in the absence of parasite transmission, with 12 differentially methylated sites in B cells and CD4+ T cells, including eight protein-coding genes. Conclusion: These genes participate in hematopoietic cell differentiation and the immune response and may be involved in immune disorders. They also have been associated with several developmental disorders and syndromes.
Collapse
Affiliation(s)
- Hans Desale
- Department of Tropical Medicine, Tulane University School of Public Health & Tropical Medicine & Tulane University Vector-Borne & Infectious Disease Research Center, New Orleans, LA 70112, USA
| | - Pierre Buekens
- Department of Epidemiology, Tulane University School of Public Health & Tropical Medicine, New Orleans, LA 70112, USA
| | - Jackeline Alger
- Instituto de Enfermedades Infecciosas y Parasitologia Antonio Vidal, Tegucigalpa, Honduras.,Ministry of Health, Hospital Escuela, Tegucigalpa, Honduras
| | - Maria Luisa Cafferata
- Unidad de Investigación Clínica y Epidemiológica Montevideo (UNICEM), Hospital de Clínicas, Montevideo, 11600, Uruguay
| | - Emily Wheeler Harville
- Department of Epidemiology, Tulane University School of Public Health & Tropical Medicine, New Orleans, LA 70112, USA
| | - Claudia Herrera
- Department of Tropical Medicine, Tulane University School of Public Health & Tropical Medicine & Tulane University Vector-Borne & Infectious Disease Research Center, New Orleans, LA 70112, USA
| | - Carine Truyens
- Laboratory of Parasitology, Faculty of Medicine, & ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles, Brussels, Belgium
| | - Eric Dumonteil
- Department of Tropical Medicine, Tulane University School of Public Health & Tropical Medicine & Tulane University Vector-Borne & Infectious Disease Research Center, New Orleans, LA 70112, USA
| |
Collapse
|
16
|
Parayiwa C, Harley D, Clark R, Behie A, Lal A. Association between severe cyclone events and birth outcomes in Queensland, Australia, 2008-2018: a population based retrospective cohort study. Aust N Z J Public Health 2022; 46:835-841. [PMID: 35735907 DOI: 10.1111/1753-6405.13273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 03/01/2022] [Accepted: 05/01/2022] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVE Investigate an association between severe tropical cyclones (TCs) and birth outcomes in an Australian population. METHODS We analysed over 600,000 singleton livebirths collected through the Queensland Perinatal Data Collection between 2008 and 2018. We estimated the odds ratios (ORs) of adverse birth outcomes using logistic multi-level modelling. RESULTS Exposure to TCs in early pregnancy was associated with significantly higher odds of preterm births in affected compared to unaffected areas during the TC year [OR=1.28, 95%CI=1.11, 1.49, p=0.001] and slightly significant higher odds in affected areas during TC years compared to non-TC years. Significantly higher odds of low birthweight births were associated with mid-pregnancy exposure to cyclone Marcia [OR=1.62, 95%CI=1.00, 2.40, p=0.016] . CONCLUSIONS Findings aligned with studies demonstrating an association between exposure to environmental stressors in early to mid-pregnancy and adverse birth outcomes. IMPLICATIONS FOR PUBLIC HEALTH There is limited research into TCs and perinatal health in Australia despite most of the population residing along coastlines and TCs presenting one of the nation's most devastating weather events. This study will inform public health practice and contribute to further research into mitigating environmental risks faced by pregnant women.
Collapse
Affiliation(s)
- Cynthia Parayiwa
- School of Archaeology and Anthropology, The Australian National University, Australian Capital Territory
| | - David Harley
- UQ Centre for Clinical Research, Faculty of Medicine, University of Queensland (UQ)
| | - Robert Clark
- Research School of Finance, Actuarial Studies and Statistics, College of Business & Economics, Australian National University, Australian Capital Territory
| | - Alison Behie
- School of Archaeology and Anthropology, The Australian National University, Australian Capital Territory
| | - Aparna Lal
- Research School of Population Health, National Centre for Epidemiology and Population Health (NCEPH), Australian National University, Australian Capital Territory
| |
Collapse
|
17
|
Using complexity science to understand the role of co-sleeping (bedsharing) in mother-infant co-regulatory processes. Infant Behav Dev 2022; 67:101723. [PMID: 35594598 DOI: 10.1016/j.infbeh.2022.101723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 04/27/2022] [Accepted: 05/04/2022] [Indexed: 11/22/2022]
Abstract
Human infants spend most of their time sleeping, but over the first few years of life their sleep becomes regulated to coincide more closely with adult sleep (Galland et al., 2012; Paavonen et al., 2020). Evidence shows that co-sleeping played a role in the evolution of infant sleep regulation, as it is part of an ancient behavioral complex representing the biopsychosocial microenvironment in which human infants co-evolved with their mothers through millions of years of human history (Ball, 2003; McKenna 1986, 1990). This paper is a conceptual, interdisciplinary, integration of the literature on mother-infant co-sleeping and other mother-infant co-regulatory processes from an evolutionary (biological) perspective, using complexity science. Viewing the mother-infant dyad as a "complex adaptive system" (CAS) shows how the CAS fits assumptions of regulatory processes and reveals the role of the CAS in the ontogeny of mother-infant co-regulation of physiological (thermoregulation, breathing, circadian rhythm coordination, nighttime synchrony, and heart rate variability) and socioemotional (attachment and cortisol activity) development.
Collapse
|
18
|
Siddeek B, Simeoni U. Epigenetics provides a bridge between early nutrition and long-term health and a target for disease prevention. Acta Paediatr 2022; 111:927-934. [PMID: 35038770 PMCID: PMC9305224 DOI: 10.1111/apa.16258] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 12/19/2022]
Abstract
Exposure to nutritional imbalance during early life can influence disease risk lifelong and across generations. In this long‐term conditioning, epigenetics constitutes a key mechanism. They bridge environmental cues and the expression of genes involved in the setting of long‐standing biological regulations in numerous organs and species. Epigenetic marks are proposed as innovative diagnostic biomarkers and potential targets in the prevention of diseases. However, a number of uncertainties make them difficult to use in clinical approaches in the context of early exposure to nutritional challenge. In conclusion, active investigations in this field are still needed before clinical applications are considered.
Collapse
Affiliation(s)
- Benazir Siddeek
- DOHaD Laboratory Division of Pediatrics Woman‐Mother‐Child‐Department Centre Hospitalier Universitaire Vaudois and University of Lausanne Lausanne Switzerland
| | - Umberto Simeoni
- DOHaD Laboratory Division of Pediatrics Woman‐Mother‐Child‐Department Centre Hospitalier Universitaire Vaudois and University of Lausanne Lausanne Switzerland
| |
Collapse
|
19
|
Genetically modified mice for research on human diseases: A triumph for Biotechnology or a work in progress? THE EUROBIOTECH JOURNAL 2022. [DOI: 10.2478/ebtj-2022-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022] Open
Abstract
Abstract
Genetically modified mice are engineered as models for human diseases. These mouse models include inbred strains, mutants, gene knockouts, gene knockins, and ‘humanized’ mice. Each mouse model is engineered to mimic a specific disease based on a theory of the genetic basis of that disease. For example, to test the amyloid theory of Alzheimer’s disease, mice with amyloid precursor protein genes are engineered, and to test the tau theory, mice with tau genes are engineered. This paper discusses the importance of mouse models in basic research, drug discovery, and translational research, and examines the question of how to define the “best” mouse model of a disease. The critiques of animal models and the caveats in translating the results from animal models to the treatment of human disease are discussed. Since many diseases are heritable, multigenic, age-related and experience-dependent, resulting from multiple gene-gene and gene-environment interactions, it will be essential to develop mouse models that reflect these genetic, epigenetic and environmental factors from a developmental perspective. Such models would provide further insight into disease emergence, progression and the ability to model two-hit and multi-hit theories of disease. The summary examines the biotechnology for creating genetically modified mice which reflect these factors and how they might be used to discover new treatments for complex human diseases such as cancers, neurodevelopmental and neurodegenerative diseases.
Collapse
|
20
|
Giannotti M, Gemignani M, Rigo P, Venuti P, De Falco S. The Role of Paternal Involvement on Behavioral Sensitive Responses and Neurobiological Activations in Fathers: A Systematic Review. Front Behav Neurosci 2022; 16:820884. [PMID: 35355925 PMCID: PMC8959913 DOI: 10.3389/fnbeh.2022.820884] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/25/2022] [Indexed: 11/30/2022] Open
Abstract
As fathering research has flourished, a growing body of studies has focused on behavioral and neurobiological mechanisms, respectively associated with caregiving sensitivity and responsiveness to infant stimuli. However, the association between these aspects and the key concept of paternal involvement in childcare (i.e., contribution in infant care in terms of time, availability, and responsibility) has been poorly investigated. The current work aims to systematically review the role of involvement in childcare on both neural activations and sensitive behaviors in fathers by examining (a) how paternal involvement has been measured and (b) whether paternal involvement has been associated with neurobiological activation and behavioral sensitive responses. Inclusion criteria were peer-reviewed quantitative studies, concerning fathers responding to infant stimuli at neurobiological or behavioral level, and including a quantitative measurement of paternal involvement in childcare. A quality rating for each study has been performed based on the measurements adopted to assess paternal involvement. Of 2,529 articles, 27 studies were included. According to our quality rating, 10 out of 27 studies included fairly good-standard measures for measuring paternal involvement, whereas 17 studies used good-standard measures. In addition, 11 studies provided details of paternal involvement in the context of neurobiological responses to infant stimuli, whereas 16 addressed paternal sensitive behaviors. Overall, only 8 studies reported relevant findings about the relationship between paternal involvement and neurobiological responses or sensitive behaviors in fathers. The present study is the first systematically evaluating the scope of paternal involvement in the field of Paternal Brain and fathers' sensitive responsiveness research. When high-standard measures are used, paternal involvement seems to play a significant role in modulating both the hormonal and the neural pathways associated with paternal behaviors. Remarkably, the role of paternal engagement may underpin an adaptive nurturance that is not dependent on pregnancy and childbirth but on caregiving experience. A promising positive link between paternal involvement and behavioral sensitivity may be expected in further studies, which will need to corroborate our conclusion by adopting detailed and appropriate measures assessing paternal involvement. As a future line of research, the inclusion of gay fathers may be beneficial for the field.
Collapse
Affiliation(s)
- Michele Giannotti
- Department of Psychology and Cognitive Sciences, University of Trento, Trento, Italy
| | - Micol Gemignani
- Department of Psychology and Cognitive Sciences, University of Trento, Trento, Italy
| | - Paola Rigo
- Department of Developmental Psychology and Socialisation, University of Padua, Padua, Italy
| | - Paola Venuti
- Department of Psychology and Cognitive Sciences, University of Trento, Trento, Italy
| | - Simona De Falco
- Department of Psychology and Cognitive Sciences, University of Trento, Trento, Italy
| |
Collapse
|
21
|
de Magalhães-Barbosa MC, Prata-Barbosa A, da Cunha AJLA. Toxic stress, epigenetics and child development. J Pediatr (Rio J) 2022; 98 Suppl 1:S13-S18. [PMID: 34793740 PMCID: PMC9510910 DOI: 10.1016/j.jped.2021.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 09/14/2021] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES To describe the concept of toxic stress, present the basics of epigenetics and discuss their relationship with child development. DATA SOURCE Narrative literature review through a search in the SciELO, Lilacs, Medline databases using the terms Adverse Childhood Experience OR Early Life Stress, Epigenomic OR Epigenetic, Child Development OR Infant Development. DATA SYNTHESIS Continuing stress response, known as toxic stress, can occur when a child experiences intense, frequent, and/or prolonged adversity-such as physical or emotional abuse, chronic neglect, for example-without adequate adult support. This toxic stress can have harmful effects on learning, behavior, and health throughout life. Epigenetics, an emerging scientific research area, shows how environmental influences affect gene expressions and explains how early experiences can impact throughout life. CONCLUSIONS Toxic stress causes changes in the human body response systems that can be explained in part by epigenetic changes, which can be temporary or long-lasting. Pediatricians must be aware of these mechanisms and their consequences, seeking to prevent them and thus promote the health, well-being, and quality of life of children, contributing to their full development.
Collapse
Affiliation(s)
| | | | - Antonio José Ledo Alves da Cunha
- Instituto D'Or de Pesquisa e Ensino (IDOR), Rio de Janeiro, RJ, Brazil; Faculdade de Medicina, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
22
|
Panisi C, Marini M. Dynamic and Systemic Perspective in Autism Spectrum Disorders: A Change of Gaze in Research Opens to A New Landscape of Needs and Solutions. Brain Sci 2022; 12:250. [PMID: 35204013 PMCID: PMC8870276 DOI: 10.3390/brainsci12020250] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 12/21/2022] Open
Abstract
The first step for a harmonious bio-psycho-social framework in approaching autism spectrum disorders (ASD) is overcoming the conflict between the biological and the psychosocial perspective. Biological research can provide clues for a correct approach to clinical practice, assuming that it would lead to the conceptualization of a pathogenetic paradigm able to account for epidemiologic and clinical findings. The upward trajectory in ASD prevalence and the systemic involvement of other organs besides the brain suggest that the epigenetic paradigm is the most plausible one. The embryo-fetal period is the crucial window of opportunity for keeping neurodevelopment on the right tracks, suggesting that women's health in pregnancy should be a priority. Maladaptive molecular pathways beginning in utero, in particular, a vicious circle between the immune response, oxidative stress/mitochondrial dysfunction, and dysbiosis-impact neurodevelopment and brain functioning across the lifespan and are the basis for progressive multisystemic disorders that account for the substantial health loss and the increased mortality in ASD. Therefore, the biological complexity of ASD and its implications for health requires the enhancement of clinical skills on these topics, to achieve an effective multi-disciplinary healthcare model. Well-balanced training courses could be a promising starting point to make a change.
Collapse
Affiliation(s)
- Cristina Panisi
- Fondazione Istituto Sacra Famiglia ONLUS, Cesano Boscone, 20090 Milan, Italy
| | - Marina Marini
- Department of Experimental, Diagnostic and Specialty Medicine, School of Medicine, University of Bologna, 40126 Bologna, Italy;
| |
Collapse
|
23
|
Panera N, Mandato C, Crudele A, Bertrando S, Vajro P, Alisi A. Genetics, epigenetics and transgenerational transmission of obesity in children. Front Endocrinol (Lausanne) 2022; 13:1006008. [PMID: 36452324 PMCID: PMC9704419 DOI: 10.3389/fendo.2022.1006008] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/27/2022] [Indexed: 11/16/2022] Open
Abstract
Sedentary lifestyle and consumption of high-calorie foods have caused a relentless increase of overweight and obesity prevalence at all ages. Its presently epidemic proportion is disquieting due to the tight relationship of obesity with metabolic syndrome and several other comorbidities which do call for urgent workarounds. The usual ineffectiveness of present therapies and failure of prevention campaigns triggered overtime a number of research studies which have unveiled some relevant aspects of obesity genetic and epigenetic inheritable profiles. These findings are revealing extremely precious mainly to serve as a likely extra arrow to allow the clinician's bow to achieve still hitherto unmet preventive goals. Evidence now exists that maternal obesity/overnutrition during pregnancy and lactation convincingly appears associated with several disorders in the offspring independently of the transmission of a purely genetic predisposition. Even the pre-conception direct exposure of either father or mother gametes to environmental factors can reprogram the epigenetic architecture of cells. Such phenomena lie behind the transfer of the obesity susceptibility to future generations through a mechanism of epigenetic inheritance. Moreover, a growing number of studies suggests that several environmental factors such as maternal malnutrition, hypoxia, and exposure to excess hormones and endocrine disruptors during pregnancy and the early postnatal period may play critical roles in programming childhood adipose tissue and obesity. A deeper understanding of how inherited genetics and epigenetics may generate an obesogenic environment at pediatric age might strengthen our knowledge about pathogenetic mechanisms and improve the clinical management of patients. Therefore, in this narrative review, we attempt to provide a general overview of the contribution of heritable genetic and epigenetic patterns to the obesity susceptibility in children, placing a particular emphasis on the mother-child dyad.
Collapse
Affiliation(s)
- Nadia Panera
- Unit of Molecular Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Claudia Mandato
- Pediatrics Section, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Salermo, Italy
- *Correspondence: Anna Alisi, ; Claudia Mandato,
| | - Annalisa Crudele
- Unit of Molecular Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Sara Bertrando
- Pediatrics Clinic, San Giovanni di Dio e Ruggi d’Aragona University Hospital, Salerno, Italy
| | - Pietro Vajro
- Pediatrics Section, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Salermo, Italy
| | - Anna Alisi
- Unit of Molecular Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- *Correspondence: Anna Alisi, ; Claudia Mandato,
| |
Collapse
|
24
|
Gombert M, Martin-Carbonell V, Pin-Arboledas G, Carrasco-Luna J, Carrasco-García Á, Codoñer-Franch P. Melatonin Levels in Children with Obesity Are Associated with Metabolic Risk and Inflammatory Parameters. Nutrients 2021; 13:nu13103629. [PMID: 34684629 PMCID: PMC8541212 DOI: 10.3390/nu13103629] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/23/2021] [Accepted: 10/13/2021] [Indexed: 12/21/2022] Open
Abstract
Melatonin, the hormone of circadian rhythm regulation, is involved in the modulation of mitochondrial activity through its antioxidant and anti-inflammatory properties. Alteration of circadian rhythms such as sleep is related to obesity and metabolic pathogenesis in adulthood, but studies during childhood are scarce. The present study investigated the association of melatonin with metabolic and inflammatory markers in children with (n = 113) and without obesity (n = 117). Melatonin was measured in saliva four and two hours before bedtime, and after one hour of sleep. Cardiometabolic factors, high sensitivity C-reactive protein, immune markers (monocyte chemoattractant protein-1, plasminogen activator inhibitor-1, tumor necrosis α and interferon-γ), leptin and ghrelin were determined. Sleep duration was recorded by a questionnaire. The melatonin level at 1 h after sleep was found to be increased more than twofold in children with obesity (90.16 (57.16–129.16) pg/mL) compared to controls (29.82 (19.05–61.54) pg/mL, p < 0.001) and was related to fat mass (rho = 0.294, p < 0.001); melatonin levels at 1 h after sleep were inversely correlated with high-density lipoprotein cholesterol. Positive correlation was found with apolipoprotein B, adipokines, high sensitivity C-reactive protein, plasminogen activator inhibitor-1 and tumor necrosis factor-α. Shorter sleep duration and earlier waking times were recorded in children with obesity. In conclusion, melatonin in children with obesity appears to be involved in the global metabolic and inflammatory alteration of this condition.
Collapse
Affiliation(s)
- Marie Gombert
- Department of Pediatrics, Obstetrics and Gynecology, Universidad de Valencia, 46010 Valencia, Spain; (M.G.); (V.M.-C.); (Á.C.-G.)
| | - Vanessa Martin-Carbonell
- Department of Pediatrics, Obstetrics and Gynecology, Universidad de Valencia, 46010 Valencia, Spain; (M.G.); (V.M.-C.); (Á.C.-G.)
| | - Gonzalo Pin-Arboledas
- Pediatric Sleep Unit, Department of Pediatrics, Quironsalud Hospital, 46010 Valencia, Spain;
| | - Joaquín Carrasco-Luna
- Department of Experimental Sciences, Universitat Católica de Valencia, 46001 Valencia, Spain;
| | - Álvaro Carrasco-García
- Department of Pediatrics, Obstetrics and Gynecology, Universidad de Valencia, 46010 Valencia, Spain; (M.G.); (V.M.-C.); (Á.C.-G.)
| | - Pilar Codoñer-Franch
- Department of Pediatrics, Obstetrics and Gynecology, Universidad de Valencia, 46010 Valencia, Spain; (M.G.); (V.M.-C.); (Á.C.-G.)
- Service of Pediatrics, University Hospital Doctor Peset, FISABIO, 46017 Valencia, Spain
- Correspondence:
| |
Collapse
|
25
|
Beleza J, Stevanović-Silva J, Coxito P, Costa RC, Ascensão A, Torrella JR, Magalhães J. Building-up fit muscles for the future: Transgenerational programming of skeletal muscle through physical exercise. Eur J Clin Invest 2021; 51:e13515. [PMID: 33580562 DOI: 10.1111/eci.13515] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/31/2021] [Accepted: 02/03/2021] [Indexed: 12/19/2022]
Abstract
'Special issue - In Utero and Early Life Programming of Aging and Disease'. Skeletal muscle (SM) adaptations to physical exercise (PE) have been extensively studied due, not only to the relevance of its in situ plasticity, but also to the SM endocrine-like effects in noncontractile tissues, such as brain, liver or adipocytes. Regular PE has been considered a pleiotropic nonpharmacological strategy to prevent and counteract the deleterious consequences of several metabolic, cardiovascular, oncological and neurodegenerative disorders. Additionally, PE performed by parents seems to have a direct impact in the offspring through the transgenerational programming of different tissues, such as SM. In fact, SM offspring programming mechanisms seems to be orchestrated, at least in part, by epigenetic machinery conditioning transcriptional or post-transcriptional processes. Ultimately, PE performed in the early in life is also a critical window of opportunity to positively modulate the juvenile and adult phenotype. Parental PE has a positive impact in several health-related offspring outcomes, such as SM metabolism, differentiation, morphology and ultimately in offspring exercise volition and endurance. Also, early-life PE counteracts conceptional-related adverse effects and induces long-lasting healthy benefits throughout adulthood. Additionally, epigenetics mechanisms seem to play a key role in the PE-induced SM adaptations. Despite the undoubtedly positive role of parental and early-life PE on SM phenotype, a strong research effort is still needed to better understand the mechanisms that positively regulate PE-induced SM programming.
Collapse
Affiliation(s)
- Jorge Beleza
- Department of Cell Biology, Physiology & Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Jelena Stevanović-Silva
- Laboratory of Metabolism and Exercise (LaMetEx), Faculty of Sport, Research Centre in Physical Activity, Health and Leisure (CIAFEL), University of Porto, Porto, Portugal
| | - Pedro Coxito
- Laboratory of Metabolism and Exercise (LaMetEx), Faculty of Sport, Research Centre in Physical Activity, Health and Leisure (CIAFEL), University of Porto, Porto, Portugal
| | - Rui Carlos Costa
- Department of Communication and Art, Research Institute for Design, Media and Culture (ID+), Aveiro University, Aveiro, Portugal
| | - António Ascensão
- Laboratory of Metabolism and Exercise (LaMetEx), Faculty of Sport, Research Centre in Physical Activity, Health and Leisure (CIAFEL), University of Porto, Porto, Portugal
| | - Joan Ramon Torrella
- Department of Cell Biology, Physiology & Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - José Magalhães
- Laboratory of Metabolism and Exercise (LaMetEx), Faculty of Sport, Research Centre in Physical Activity, Health and Leisure (CIAFEL), University of Porto, Porto, Portugal
| |
Collapse
|
26
|
Grumi S, Provenzi L, Accorsi P, Biasucci G, Cavallini A, Decembrino L, Falcone R, Fazzi EM, Gardella B, Giacchero R, Guerini P, Grossi E, Magnani ML, Mariani EM, Nacinovich R, Pantaleo D, Pisoni C, Prefumo F, Sabatini C, Scelsa B, Spartà MV, Spinillo A, Giorda R, Orcesi S, Borgatti R. Depression and Anxiety in Mothers Who Were Pregnant During the COVID-19 Outbreak in Northern Italy: The Role of Pandemic-Related Emotional Stress and Perceived Social Support. Front Psychiatry 2021; 12:716488. [PMID: 34539466 PMCID: PMC8446509 DOI: 10.3389/fpsyt.2021.716488] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/09/2021] [Indexed: 12/28/2022] Open
Abstract
The COVID-19 pandemic is a collective trauma that is threatening citizens' mental health resulting in increased emotional stress, reduced social support, and heightened risk for affective symptoms. The present study aimed to investigate the effects of antenatal pandemic-related emotional stress and perceived social support on the symptoms of depression and anxiety of mothers who were pregnant during the initial COVID-19 outbreak in northern Italy. A sample of 281 mothers was enrolled at eight maternity units in the first hotspot region of the COVID-19 outbreak in northern Italy. Participants filled out online questionnaires assessing the direct or indirect exposure to the SARS-CoV-2 virus, pandemic-related stress, perceived social support, as well as symptoms of depression and anxiety. Depressive and anxious symptomatology was above clinical concern, respectively, in 26 and 32% of the respondents. Mothers who reported no exposure to SARS-CoV-2 during pregnancy and those who reported at least one direct or indirect exposure did not differ in terms of affective symptoms. Continuous scores and risk for severe depression and anxiety were positively associated with prenatal pandemic-related emotional stress and negatively linked with perceived social support during pregnancy. Women who become mothers during the COVID-19 emergency may be at high risk for affective problems. Dedicated preventive programs are needed to provide adequate preventive support and care for maternal mental health during and after the COVID-19 pandemic.
Collapse
Affiliation(s)
- Serena Grumi
- Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Livio Provenzi
- Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Patrizia Accorsi
- Unit of Child and Adolescence Neuropsychiatry, ASST Spedali Civili, Brescia, Italy
| | - Giacomo Biasucci
- Pediatrics & Neonatology Unit, Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Anna Cavallini
- Child and Adolescent Mental Health, San Gerardo Hospital, Monza, Italy
| | | | | | - Elisa Maria Fazzi
- Unit of Child and Adolescence Neuropsychiatry, ASST Spedali Civili, Brescia, Italy
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Barbara Gardella
- Department of Obstetrics and Gynecology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | | | - Paola Guerini
- Pediatric Unit and Neonatal Unit, ASST Pavia, Pavia, Italy
| | - Elena Grossi
- Pediatrics & Neonatology Unit, Guglielmo da Saliceto Hospital, Piacenza, Italy
| | | | | | - Renata Nacinovich
- Child and Adolescent Mental Health, San Gerardo Hospital, Monza, Italy
- School of Medicine and Surgery and Milan Center for Neuroscience, Università Bicocca, Milan, Italy
| | - Dario Pantaleo
- Pediatric Unit and Neonatal Unit, ASST Pavia, Pavia, Italy
| | - Camilla Pisoni
- Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Federico Prefumo
- Unit of Child and Adolescence Neuropsychiatry, ASST Spedali Civili, Brescia, Italy
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | | | - Barbara Scelsa
- Pediatric Neurology Unit, V. Buzzi Children's Hospital, Milan, Italy
| | | | - Arsenio Spinillo
- Department of Obstetrics and Gynecology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Roberto Giorda
- Biology Lab, Scientific Institute IRCCS E. Medea, Bosisio Parini, Italy
| | - Simona Orcesi
- Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Renato Borgatti
- Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
27
|
Lagercrantz H. Young Investigator Award 2020. Acta Paediatr 2021; 110:2492. [PMID: 33837567 DOI: 10.1111/apa.15865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 03/30/2021] [Indexed: 11/28/2022]
|
28
|
Akbari B, Ghahri-Saremi N, Soltantoyeh T, Hadjati J, Ghassemi S, Mirzaei HR. Epigenetic strategies to boost CAR T cell therapy. Mol Ther 2021; 29:2640-2659. [PMID: 34365035 DOI: 10.1016/j.ymthe.2021.08.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 07/19/2021] [Accepted: 07/31/2021] [Indexed: 02/08/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has led to a paradigm shift in cancer immunotherapy, but still several obstacles limit CAR T cell efficacy in cancers. Advances in high-throughput technologies revealed new insights into the role that epigenetic reprogramming plays in T cells. Mechanistic studies as well as comprehensive epigenome maps revealed an important role for epigenetic remodeling in T cell differentiation. These modifications shape the overall immune response through alterations in T cell phenotype and function. Here, we outline how epigenetic modifications in CAR T cells can overcome barriers limiting CAR T cell effectiveness, particularly in immunosuppressive tumor microenvironments. We also offer our perspective on how selected epigenetic modifications can boost CAR T cells to ultimately improve the efficacy of CAR T cell therapy.
Collapse
Affiliation(s)
- Behnia Akbari
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran
| | - Navid Ghahri-Saremi
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran
| | - Tahereh Soltantoyeh
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran
| | - Jamshid Hadjati
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran
| | - Saba Ghassemi
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran.
| |
Collapse
|
29
|
Fornari E, Brusati M, Maffeis C. Nutritional Strategies for Childhood Obesity Prevention. Life (Basel) 2021; 11:532. [PMID: 34201017 PMCID: PMC8227398 DOI: 10.3390/life11060532] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/31/2021] [Accepted: 05/31/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Reducing the spread of obesity represents a challenge for clinicians in which obesity prevention plays a key role in achieving this purpose. The aim of this review is to analyze the nutritional interventions that can be implemented to prevent childhood obesity. METHODS Searching PubMed and Cochrane Library between 2019 and 2021. Further searching with no date range for articles selected for their specific relevance in the pediatric area or for their scientific relevance. A total of 871 articles were identified and 90 were included. RESULTS We organized the results of the selected articles into age groups, and according to the subjects targeted for interventions or to the site of interventions, reserving an in-depth analysis on specific nutritional aspects. Promotion of breastfeeding, reduction of protein content of formulated milks, and diet of the first 12-24 months, involving family and schools in interventions that promote physical activity and healthy diet, are promising strategies for reduction of the risk of obesity. To increase the efficacy of interventions, a multidimensional approach is crucial. CONCLUSIONS A multidimensional approach, which takes into consideration different areas of intervention, is pivotal for childhood obesity prevention. Integrated programs involving several components (nutrition and physical activity at first) at different levels (individual, family, school, and institutional) are crucial.
Collapse
Affiliation(s)
- Elena Fornari
- Department of Surgery, Dentistry, Pediatrics and Gynecology, Section of Pediatric Diabetes and Metabolism, University and Azienda Ospedaliera Universitaria Integrata of Verona, 37126 Verona, Italy;
| | - Marco Brusati
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy;
| | - Claudio Maffeis
- Department of Surgery, Dentistry, Pediatrics and Gynecology, Section of Pediatric Diabetes and Metabolism, University and Azienda Ospedaliera Universitaria Integrata of Verona, 37126 Verona, Italy;
| |
Collapse
|
30
|
Dieckmann L, Lahti-Pulkkinen M, Kvist T, Lahti J, DeWitt PE, Cruceanu C, Laivuori H, Sammallahti S, Villa PM, Suomalainen-König S, Eriksson JG, Kajantie E, Raikkönen K, Binder EB, Czamara D. Characteristics of epigenetic aging across gestational and perinatal tissues. Clin Epigenetics 2021; 13:97. [PMID: 33926514 PMCID: PMC8082803 DOI: 10.1186/s13148-021-01080-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/14/2021] [Indexed: 12/11/2022] Open
Abstract
Background Epigenetic clocks have been used to indicate differences in biological states between individuals of same chronological age. However, so far, only few studies have examined epigenetic aging in newborns—especially regarding different gestational or perinatal tissues. In this study, we investigated which birth- and pregnancy-related variables are most important in predicting gestational epigenetic age acceleration or deceleration (i.e., the deviation between gestational epigenetic age estimated from the DNA methylome and chronological gestational age) in chorionic villus, placenta and cord blood tissues from two independent study cohorts (ITU, n = 639 and PREDO, n = 966). We further characterized the correspondence of epigenetic age deviations between these tissues. Results Among the most predictive factors of epigenetic age deviations in single tissues were child sex, birth length, maternal smoking during pregnancy, maternal mental disorders until childbirth, delivery mode and parity. However, the specific factors related to epigenetic age deviation and the direction of association differed across tissues. In individuals with samples available from more than one tissue, relative epigenetic age deviations were not correlated across tissues. Conclusion Gestational epigenetic age acceleration or deceleration was not related to more favorable or unfavorable factors in one direction in the investigated tissues, and the relative epigenetic age differed between tissues of the same person. This indicates that epigenetic age deviations associate with distinct, tissue specific, factors during the gestational and perinatal period. Our findings suggest that the epigenetic age of the newborn should be seen as a characteristic of a specific tissue, and less as a general characteristic of the child itself. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-021-01080-y.
Collapse
Affiliation(s)
- Linda Dieckmann
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, München, Germany.,International Max Planck Research School for Translational Psychiatry, München, Germany
| | - Marius Lahti-Pulkkinen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,National Institute for Health and Welfare, Helsinki, Finland.,Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Tuomas Kvist
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jari Lahti
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Peter E DeWitt
- Section of Informatics and Data Science, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Cristiana Cruceanu
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, München, Germany
| | - Hannele Laivuori
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Human Genetics, Helsinki, Finland.,Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Department of Obstetrics and Gynecology- Faculty of Medicine and Health Technology, Tampere University Hospital and Tampere University, Tampere, Finland
| | - Sara Sammallahti
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,National Institute for Health and Welfare, Helsinki, Finland.,Children's Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland.,Department of Child and Adolescent Psychiatry, Erasmus MC, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Pia M Villa
- Department of Obstetrics and Gynecology- Faculty of Medicine and Health Technology, Tampere University Hospital and Tampere University, Tampere, Finland.,Department of Obstetrics and Gynecology, Helsinki University Central Hospital, Helsinki, Finland.,Hyvinkää Hospital, Helsinki and Uusimaa Hospital District, Hyvinkää, Finland
| | - Sanna Suomalainen-König
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Johan G Eriksson
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Folkhälsan Research Center, Helsinki, Finland.,Department of Obstetrics & Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Eero Kajantie
- National Institute for Health and Welfare, Helsinki, Finland.,Children's Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland.,Faculty of Medicine, PEDEGO Research Unit, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.,Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Katri Raikkönen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Elisabeth B Binder
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, München, Germany.,Department of Psychiatry and Behavioral Sciences, School of Medicine, Emory University, Atlanta, GA, USA
| | - Darina Czamara
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, München, Germany.
| |
Collapse
|
31
|
Čater M, Majdič G. How early maternal deprivation changes the brain and behavior? Eur J Neurosci 2021; 55:2058-2075. [PMID: 33870558 DOI: 10.1111/ejn.15238] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 04/06/2021] [Accepted: 04/09/2021] [Indexed: 01/30/2023]
Abstract
Early life stress can adversely influence brain development and reprogram brain function and consequently behavior in adult life. Adequate maternal care in early childhood is therefore particularly important for the normal brain development, and adverse early life experiences can lead to altered emotional, behavioral, and neuroendocrine stress responses in the adulthood. As a form of neonatal stress, maternal deprivation/separation is often used in behavioral studies to examine the effects of early life stress and for modeling the development of certain psychiatric disorders and brain pathologies in animal models. The temporary loss of maternal care during the critical postpartum periods remodels the offspring's brain and provokes long-term effects on learning and cognition, the development of mental disorders, aggression, and an increased tendency for the drug abuse. Early life stress through maternal deprivation affects neuroendocrine responses to stress in adolescence and adulthood by dysregulating the hypothalamic-pituitary-adrenal axis and permanently disrupts stress resilience. In this review, we focused on how improper maternal care during early postnatal life affects brain development resulting in modified behavior later in life.
Collapse
Affiliation(s)
- Maša Čater
- Veterinary Faculty, Laboratory for Animal Genomics, Institute for Preclinical Studies, University of Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, Institute of Physiology, University of Maribor, Maribor, Slovenia
| | - Gregor Majdič
- Veterinary Faculty, Laboratory for Animal Genomics, Institute for Preclinical Studies, University of Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, Institute of Physiology, University of Maribor, Maribor, Slovenia
| |
Collapse
|
32
|
Berretta E, Guida E, Forni D, Provenzi L. Glucocorticoid receptor gene (NR3C1) methylation during the first thousand days: Environmental exposures and developmental outcomes. Neurosci Biobehav Rev 2021; 125:493-502. [PMID: 33689802 DOI: 10.1016/j.neubiorev.2021.03.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 10/07/2020] [Accepted: 03/03/2021] [Indexed: 02/08/2023]
Abstract
The first 1000 days from conception are a sensitive period for human development programming. During this period, environmental exposures may result in long-lasting epigenetic imprints that contribute to future developmental trajectories. The present review reports on the effects of adverse and protective environmental conditions occurring during the first 1000 days on glucocorticoid receptor gene (NR3C1) regulation in humans. Thirty-four studies were included. Wide variations emerged for biological tissues, number and position of analyzed CpG sites, and age at methylation and outcomes assessment. Increased NR3C1 methylation associated with first 1000 days stress exposures. Maternal caregiving behaviors significantly buffered precocious stress exposures. A less robust pattern of findings emerged for the association of NR3C1 methylation with physical health, neurobehavioral and neuroendocrine outcomes. Although drawing comprehensive conclusions is partially hindered by methodological limitations, the present review underlines the relevance of the first 1000 days from conception as a time window for developmental plasticity. Prospective cohort studies and epigenome-wide approaches may increase our understanding of dynamics epigenetic changes and their consequences for child development.
Collapse
Affiliation(s)
- Erica Berretta
- Experimental and Behavioral Neurophysiology Lab, Scientific Institute IRCCS Fondazione Santa Lucia, Roma, Italy
| | - Elena Guida
- 0-3 Center for the At-Risk Infant, Scientific Institute IRCCS E. Medea, Bosisio Parini, Italy
| | - Diego Forni
- Bioinformatics, Scientific Institute IRCCS E. Medea, Bosisio Parini, Italy
| | - Livio Provenzi
- Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy.
| |
Collapse
|
33
|
Fan XY, Shi G, Zhao P. Neonatal Sevoflurane Exposure Impairs Learning and Memory by the Hypermethylation of Hippocampal Synaptic Genes. Mol Neurobiol 2021; 58:895-904. [PMID: 33052583 DOI: 10.1007/s12035-020-02161-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/04/2020] [Indexed: 12/20/2022]
Abstract
Sevoflurane anesthesia is widely used in pediatric patients. Clinical studies report memory impairment in those exposed to general anesthesia early in life. DNA methylation is essential for the modulation of synaptic plasticity through regulating the transcription of synaptic genes. Therefore, we tested whether neonatal sevoflurane exposure affects learning and memory underlying the hippocampal DNA methylation of synaptic genes. Male Sprague-Dawley rats were exposed to 3% sevoflurane or air for 2 h daily from postnatal day 7 (P7) to P9. 5-aza-2-deoxycytidine (5-AZA), an inhibitor of DNA methyltransferases (DNMTs), was intraperitoneally injected 30 min before sevoflurane or air exposure on P7-9. The rats were euthanized 6, 12, 24 h, and 28 days after the last sevoflurane exposure, followed by the determination of global and gene-specific DNA methylation. The expression of synaptic proteins and synaptic density and the transcription of Dnmts and ten eleven translocations (Tets) in the hippocampus were measured. The ability of learning and memory was assessed using Morris water maze, novel object recognition, and intruder tests. Repeated neonatal sevoflurane exposure impaired cognitive, social, and spatial memory. The memory impairment was associated with the increased Dnmt1, Dnmt3a, and 5-methylcytosine level and the decreased Tet1 and 5-hydromethylcytosine level. Sevoflurane subsequently induced hypermethylation of Shank2, Psd95, Syn1, and Syp gene and down-regulated the expression of synaptic proteins, which finally led to the decrease of synaptic density in a time-dependent manner. Notably, 5-AZA pretreatment ameliorated learning and memory in sevoflurane-treated rats. In conclusion, neonatal exposure to sevoflurane can impair learning and memory through DNA methylation of synaptic genes.
Collapse
Affiliation(s)
- Xin-Yu Fan
- Department of Anesthesiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, China
| | - Guang Shi
- Department of Neurology, Liaoning Provincial People's Hospital, Shenyang, China
| | - Ping Zhao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, China.
| |
Collapse
|
34
|
Panisi C, Guerini FR, Abruzzo PM, Balzola F, Biava PM, Bolotta A, Brunero M, Burgio E, Chiara A, Clerici M, Croce L, Ferreri C, Giovannini N, Ghezzo A, Grossi E, Keller R, Manzotti A, Marini M, Migliore L, Moderato L, Moscone D, Mussap M, Parmeggiani A, Pasin V, Perotti M, Piras C, Saresella M, Stoccoro A, Toso T, Vacca RA, Vagni D, Vendemmia S, Villa L, Politi P, Fanos V. Autism Spectrum Disorder from the Womb to Adulthood: Suggestions for a Paradigm Shift. J Pers Med 2021; 11:70. [PMID: 33504019 PMCID: PMC7912683 DOI: 10.3390/jpm11020070] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 01/10/2021] [Accepted: 01/19/2021] [Indexed: 02/07/2023] Open
Abstract
The wide spectrum of unique needs and strengths of Autism Spectrum Disorders (ASD) is a challenge for the worldwide healthcare system. With the plethora of information from research, a common thread is required to conceptualize an exhaustive pathogenetic paradigm. The epidemiological and clinical findings in ASD cannot be explained by the traditional linear genetic model, hence the need to move towards a more fluid conception, integrating genetics, environment, and epigenetics as a whole. The embryo-fetal period and the first two years of life (the so-called 'First 1000 Days') are the crucial time window for neurodevelopment. In particular, the interplay and the vicious loop between immune activation, gut dysbiosis, and mitochondrial impairment/oxidative stress significantly affects neurodevelopment during pregnancy and undermines the health of ASD people throughout life. Consequently, the most effective intervention in ASD is expected by primary prevention aimed at pregnancy and at early control of the main effector molecular pathways. We will reason here on a comprehensive and exhaustive pathogenetic paradigm in ASD, viewed not just as a theoretical issue, but as a tool to provide suggestions for effective preventive strategies and personalized, dynamic (from womb to adulthood), systemic, and interdisciplinary healthcare approach.
Collapse
Affiliation(s)
- Cristina Panisi
- Fondazione Istituto Sacra Famiglia ONLUS, Cesano Boscone, 20090 Milan, Italy;
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy;
| | - Franca Rosa Guerini
- IRCCS Fondazione Don Carlo Gnocchi, ONLUS, 20148 Milan, Italy; (M.C.); (M.S.)
| | | | - Federico Balzola
- Division of Gastroenterology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Turin, 10126 Turin, Italy;
| | - Pier Mario Biava
- Scientific Institute of Research and Care Multimedica, 20138 Milan, Italy;
| | - Alessandra Bolotta
- DIMES, School of Medicine, University of Bologna, 40126 Bologna, Italy; (P.M.A.); (A.B.); (A.G.)
| | - Marco Brunero
- Department of Pediatric Surgery, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Ernesto Burgio
- ECERI—European Cancer and Environment Research Institute, Square de Meeus 38-40, 1000 Bruxelles, Belgium;
| | - Alberto Chiara
- Dipartimento Materno Infantile ASST, 27100 Pavia, Italy;
| | - Mario Clerici
- IRCCS Fondazione Don Carlo Gnocchi, ONLUS, 20148 Milan, Italy; (M.C.); (M.S.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
| | - Luigi Croce
- Centro Domino per l’Autismo, Universita’ Cattolica Brescia, 20139 Milan, Italy;
| | - Carla Ferreri
- National Research Council of Italy, Institute of Organic Synthesis and Photoreactivity (ISOF), 40129 Bologna, Italy;
| | - Niccolò Giovannini
- Department of Obstetrics and Gynecology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Alessandro Ghezzo
- DIMES, School of Medicine, University of Bologna, 40126 Bologna, Italy; (P.M.A.); (A.B.); (A.G.)
| | - Enzo Grossi
- Autism Research Unit, Villa Santa Maria Foundation, 22038 Tavernerio, Italy;
| | - Roberto Keller
- Adult Autism Centre DSM ASL Città di Torino, 10138 Turin, Italy;
| | - Andrea Manzotti
- RAISE Lab, Foundation COME Collaboration, 65121 Pescara, Italy;
| | - Marina Marini
- DIMES, School of Medicine, University of Bologna, 40126 Bologna, Italy; (P.M.A.); (A.B.); (A.G.)
| | - Lucia Migliore
- Medical Genetics Laboratories, Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy; (L.M.); (A.S.)
| | - Lucio Moderato
- Fondazione Istituto Sacra Famiglia ONLUS, Cesano Boscone, 20090 Milan, Italy;
| | - Davide Moscone
- Associazione Spazio Asperger ONLUS, Centro Clinico CuoreMenteLab, 00141 Rome, Italy;
| | - Michele Mussap
- Neonatal Intensive Care Unit, Department of Surgical Sciences, Puericulture Institute and Neonatal Section, Azienda Ospedaliera Universitaria, 09100 Cagliari, Italy; (M.M.); (V.F.)
| | - Antonia Parmeggiani
- Child Neurology and Psychiatry Unit, IRCCS ISNB, S. Orsola-Malpighi Hospital, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy;
| | - Valentina Pasin
- Milan Institute for health Care and Advanced Learning, 20124 Milano, Italy;
| | | | - Cristina Piras
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy;
| | - Marina Saresella
- IRCCS Fondazione Don Carlo Gnocchi, ONLUS, 20148 Milan, Italy; (M.C.); (M.S.)
| | - Andrea Stoccoro
- Medical Genetics Laboratories, Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy; (L.M.); (A.S.)
| | - Tiziana Toso
- Unione Italiana Lotta alla Distrofia Muscolare UILDM, 35100 Padova, Italy;
| | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council of Italy, 70126 Bari, Italy;
| | - David Vagni
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy, 98164 Messina, Italy;
| | | | - Laura Villa
- Scientific Institute, IRCCS Eugenio Medea, Via Don Luigi Monza 20, 23842 Bosisio Parini, Italy;
| | - Pierluigi Politi
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy;
| | - Vassilios Fanos
- Neonatal Intensive Care Unit, Department of Surgical Sciences, Puericulture Institute and Neonatal Section, Azienda Ospedaliera Universitaria, 09100 Cagliari, Italy; (M.M.); (V.F.)
- Neonatal Intensive Care Unit, Azienda Ospedaliera Universitaria, 09042 Cagliari, Italy
| |
Collapse
|
35
|
Provenzi L, Grumi S, Giorda R, Biasucci G, Bonini R, Cavallini A, Decembrino L, Drera B, Falcone R, Fazzi E, Gardella B, Giacchero R, Nacinovich R, Pisoni C, Prefumo F, Scelsa B, Spartà MV, Veggiotti P, Orcesi S, Borgatti R. Measuring the Outcomes of Maternal COVID-19-related Prenatal Exposure (MOM-COPE): study protocol for a multicentric longitudinal project. BMJ Open 2020; 10:e044585. [PMID: 33384402 PMCID: PMC7780424 DOI: 10.1136/bmjopen-2020-044585] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION COVID-19 is a highly infectious respiratory disease that rapidly emerged as an unprecedented epidemic in Europe, with a primary hotspot in Northern Italy during the first months of 2020. Its high infection rate and rapid spread contribute to set the risk for relevant psychological stress in citizens. In this context, mother-infant health is at risk not only because of potential direct exposure to the virus but also due to high levels of stress experienced by mothers from conception to delivery. Prenatal stress exposure associates with less-than-optimal child developmental outcomes, and specific epigenetic mechanisms (eg, DNA methylation) may play a critical role in mediating this programming association. METHODS AND ANALYSIS We present the methodological protocol for a longitudinal, multicentric study on the behavioural and epigenetic effects of COVID-19-related prenatal stress in a cohort of mother-infant dyads in Northern Italy. The dyads will be enrolled at 10 facilities in Northern Italy. Saliva samples will be collected at birth to assess the methylation status of specific genes linked with stress regulation in mothers and newborns. Mothers will provide retrospective data on COVID-19-related stress during pregnancy. At 3, 6 and 12 months, mothers will provide data on child behavioural and socioemotional outcomes, their own psychological status (stress, depressive and anxious symptoms) and coping strategies. At 12 months, infants and mothers will be videotaped during semistructured interaction to assess maternal sensitivity and infant's relational functioning. ETHICS AND DISSEMINATION This study was approved by the Ethics Committee (Pavia). Results will be published in peer-reviewed journals and presented at national and international scientific conferences. TRIAL REGISTRATION NUMBER NCT04540029; Pre-results.
Collapse
Affiliation(s)
- Livio Provenzi
- Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Serena Grumi
- Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Roberto Giorda
- Biology Lab, Scientific Institute, IRCCS E. Medea, Bosisi Parini, Italy
| | - Giacomo Biasucci
- Pediatrics & Neonatology Unit, Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Renza Bonini
- Pediatrics & Neonatology Unit, Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Anna Cavallini
- Child and Adolescent Mental Health, San Gerardo Hospital, Monza, Italy
| | - Lidia Decembrino
- Pediatric Unit and Neonatal Unit, Ospedale Civile di Vigevano, ASST di Pavia, Vigevano, Italy
| | - Bruno Drera
- Neonatal Intensive Care Unit, Azienda Istituti Ospitalieri di Cremona, Cremona, Italy
| | - Rossana Falcone
- Pediatric Unit and Neonatal Unit, Ospedale Civile di Vigevano, ASST di Pavia, Vigevano, Italy
| | - Elisa Fazzi
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Unit of Child and Adolescence Neuropsychiatry, Azienda Ospedaliera Spedali Civili di Brescia, Brescia, Italy
| | - Barbara Gardella
- Department of Obstetrics and Gynecology, IRCCS Foundation Policlinico San Matteo, Pavia, Italy
| | | | - Renata Nacinovich
- Child and Adolescent Mental Health, San Gerardo Hospital, Monza, Italy
- School of Medicine and Surgery and Milan Center for Neuroscience, University of Milano Bicocca, Milano, Italy
| | - Camilla Pisoni
- Neonatal Intensive Care Unit, IRCCS Foundation Policlinico San Matteo, Pavia, Italy
| | - Federico Prefumo
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Barbara Scelsa
- Unit of Pediatric Neurology, Buzzi Children's Hospital, Milano, Italy
| | | | - Pierangelo Veggiotti
- Unit of Pediatric Neurology, Buzzi Children's Hospital, Milano, Italy
- Biomedical and Clinical Science Department, University of Milano, Milano, Italy
| | - Simona Orcesi
- Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Renato Borgatti
- Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
36
|
A global perspective on parental stress in the neonatal intensive care unit: a meta-analytic study. J Perinatol 2020; 40:1739-1752. [PMID: 32901116 DOI: 10.1038/s41372-020-00798-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 08/15/2020] [Accepted: 08/20/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVES The Parental Stressor Scale: Neonatal Intensive Care Unit (PSS:NICU) is a well-validated tool to assess different sources of stress in parents during the NICU hospitalization of their infant. The present meta-analytic study assessed the relative impact of different NICU-related sources of parental stress in a pool of studies conducted in a wide set of different countries. Also, differences in stress levels by parent gender and country, as well as the impact of infants' neonatal characteristics and clinical conditions were explored. METHODS Records were searched on PubMed, Scopus, and Web of Science (January 1993-December 2019). A purposive open search string was adopted: ["PSS:NICU"] OR ["PSS-NICU"] OR ["Parental Stressor Scale"]. A multiple random-effect meta-analysis was conducted on data from 53 studies extracted by independent coders. RESULTS Parental role alteration emerged as the greatest source of stress for both mothers and fathers. Mothers reported higher stress levels compared to fathers. A significant difference emerged only for the subscale related to sights and sounds physical stimuli. No significant effects of infants' neonatal characteristics (gestational age, birth weight) and clinical conditions (comorbidities) emerged. A marginal positive effect of NICU length of stay emerged on the global level of parents' stress. CONCLUSIONS The current meta-analysis underlines that parental stress related to NICU admission is a worldwide healthcare issue. Immediate and tailored support to parents after the birth of their at-risk infant should be prioritized to reduce parental stress and to promote mothers and fathers' emotional well-being and new-born neurodevelopmental outcomes.
Collapse
|
37
|
Ozkan H, Tuzun F, Taheri S, Korhan P, Akokay P, Yılmaz O, Duman N, Özer E, Tufan E, Kumral A, Özkul Y. Epigenetic Programming Through Breast Milk and Its Impact on Milk-Siblings Mating. Front Genet 2020; 11:569232. [PMID: 33133155 PMCID: PMC7565666 DOI: 10.3389/fgene.2020.569232] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/01/2020] [Indexed: 12/19/2022] Open
Abstract
Background The epigenetic effects of transmission of certain regulatory molecules, such as miRNAs, through maternal milk on future generations, are still unknown and have not been fully understood yet. We hypothesized that breastfeeding regularly by adoptive-mother may cause transmission of miRNAs as epigenetic regulating factors to the infant, and the marriage of milk-siblings may cause various pathologies in the future generations. Results A cross-fostering model using a/a and Avy/a mice had been established. F2 milk-sibling and F2 control groups were obtained from mating of milk-siblings or unrelated mice. Randomized selected animals in the both F2 groups were sacrificed for miRNA expression studies and the remainings were followed for phenotypic changes (coat color, obesity, hyperglycemia, liver pathology, and life span). The lifespan in the F2 milk-sibling group was shorter than the control group (387 vs 590 days, p = 0.011) and they were more obese during the aging period. Histopathological examination of liver tissues revealed abnormal findings in F2 milk-sibling group. In order to understand the epigenetic mechanisms leading to these phenotypic changes, we analyzed miRNA expression differences between offspring of milk-sibling and control matings and focused on the signaling pathways regulating lifespan and metabolism. Bioinformatic analysis demonstrated that differentially expressed miRNAs were associated with pathways regulating metabolism, survival, and cancer development such as the PI3K-Akt, ErbB, mTOR, and MAPK, insulin signaling pathways. We further analyzed the expression patterns of miR-186-5p, miR-141-3p, miR-345-5p, and miR-34c-5p and their candidate target genes Mapk8, Gsk3b, and Ppargc1a in ovarian and liver tissues. Conclusion Our findings support for the first time that the factors modifying the epigenetic mechanisms may be transmitted by breast milk and these epigenetic interactions may be transferred transgenerationally. Results also suggested hereditary epigenetic effects of cross-fostering on future generations and the impact of mother-infant dyad on epigenetic programming.
Collapse
Affiliation(s)
- Hasan Ozkan
- Department of Pediatrics, Division of Neonatology, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - Funda Tuzun
- Department of Pediatrics, Division of Neonatology, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - Serpil Taheri
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, Turkey.,Erciyes University's Betul-Ziya Eren Genome and Stem Cell (Genkok), Kayseri, Turkey
| | - Peyda Korhan
- İzmir Biomedicine and Genome Center, Dokuz Eylul University, İzmir, Turkey
| | - Pınar Akokay
- Department of Medical Laboratory Techniques, İzmir Kavram University, İzmir, Turkey
| | - Osman Yılmaz
- The Experimental Animal Laboratory, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - Nuray Duman
- Department of Pediatrics, Division of Neonatology, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - Erdener Özer
- Department of Pathology, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - Esra Tufan
- Erciyes University's Betul-Ziya Eren Genome and Stem Cell (Genkok), Kayseri, Turkey
| | - Abdullah Kumral
- Department of Pediatrics, Division of Neonatology, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - Yusuf Özkul
- Erciyes University's Betul-Ziya Eren Genome and Stem Cell (Genkok), Kayseri, Turkey.,Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| |
Collapse
|
38
|
Linnér A, Westrup B, Lode-Kolz K, Klemming S, Lillieskold S, Markhus Pike H, Morgan B, Bergman NJ, Rettedal S, Jonas W. Immediate parent-infant skin-to-skin study (IPISTOSS): study protocol of a randomised controlled trial on very preterm infants cared for in skin-to-skin contact immediately after birth and potential physiological, epigenetic, psychological and neurodevelopmental consequences. BMJ Open 2020; 10:e038938. [PMID: 32636292 PMCID: PMC7342825 DOI: 10.1136/bmjopen-2020-038938] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
INTRODUCTION In Scandinavia, 6% of infants are born preterm, before 37 gestational weeks. Instead of continuing in the in-utero environment, maturation needs to occur in a neonatal unit with support of vital functions, separated from the mother's warmth, nutrition and other benefits. Preterm infants face health and neurodevelopment challenges that may also affect the family and society at large. There is evidence of benefit from immediate and continued skin-to-skin contact (SSC) for term and moderately preterm infants and their parents but there is a knowledge gap on its effect on unstable very preterm infants when initiated immediately after birth. METHODS AND ANALYSIS In this ongoing randomised controlled trial from Stavanger, Norway and Stockholm, Sweden, we are studying 150 infants born at 28+0 to 32+6 gestational weeks, randomised to receive care immediately after birth in SSC with a parent or conventionally in an incubator. The primary outcome is cardiorespiratory stability according to the stability of the cardiorespiratory system in the preterm score. Secondary outcomes are autonomic stability, thermal control, infection control, SSC time, breastfeeding and growth, epigenetic profile, microbiome profile, infant behaviour, stress resilience, sleep integrity, cortical maturation, neurodevelopment, mother-infant attachment and attunement, and parent experience and mental health. ETHICS AND DISSEMINATION The study has ethical approval from the Swedish Ethical Review Authority (2017/1135-31/3, 2019-03361) and the Norwegian Regional Ethical Committee (2015/889). The study is conducted according to good clinical practice and the Helsinki declaration. The results of the study will increase the knowledge about the mechanisms behind the effects of SSC for very preterm infants by dissemination to the scientific community through articles and at conferences, and to the society through parenting classes and magazines. STUDY STATUS Recruiting since April 2018. Expected trial termination June 2021. TRIAL REGISTRATION NUMBER NCT03521310 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Agnes Linnér
- Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
- Neonatal Unit, Karolinska University Hospital, Stockholm, Sweden
| | - Björn Westrup
- Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Karoline Lode-Kolz
- Department of Paediatrics, Stavanger Universitetssjukehus, Stavanger, Norway
| | - Stina Klemming
- Neonatal Unit, Karolinska University Hospital, Stockholm, Sweden
| | - Siri Lillieskold
- Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
- Neonatal Unit, Karolinska University Hospital, Stockholm, Sweden
| | - Hanne Markhus Pike
- Department of Paediatrics, Stavanger Universitetssjukehus, Stavanger, Norway
| | - Barak Morgan
- Global Risk Governance Programme, Law Faculty, University of Cape Town, Rondebosch, Western Cape, South Africa
- NRF Centre of Excellence in Human Development, University of the Witwatersrand, Johannesburg-Braamfontein, Gauteng, South Africa
| | | | - Siren Rettedal
- Department of Paediatrics, Stavanger Universitetssjukehus, Stavanger, Norway
| | - Wibke Jonas
- Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
39
|
|